{"title":"IGLC1 is an independent prognostic marker and potent therapeutic target in osteosarcoma.","authors":"Zhenjie Wu, Xinlian Xie, Guangfu Shi, Kebin Ning, Jinmin Zhao","doi":"10.1007/s12672-025-02653-6","DOIUrl":"https://doi.org/10.1007/s12672-025-02653-6","url":null,"abstract":"<p><strong>Background: </strong>Immunoglobulin lambda constant 1 (IGLC1) has been implicated in cancer progression, but its role in osteosarcoma remains unclear. This study endeavored to explore the levels of IGLC1 expression, its implications for patient prognosis, and its functional impact within the context of osteosarcoma.</p><p><strong>Methods: </strong>IGLC1 expression was analyzed in 108 osteosarcoma tissues and 42 non-neoplastic bone samples. Associations with clinicopathological features were evaluated using chi-square tests. Prognostic value was assessed via univariate and multivariate Cox regression analyses. Functional assays, including CCK-8, colony formation, transwell migration, wound healing, and invasion assays, were conducted following IGLC1 knockdown in MG-63 and 143B cell lines.</p><p><strong>Results: </strong>High IGLC1 expression was observed in 55.6% of osteosarcoma cases, significantly higher than in non-neoplastic bone tissues (P < 0.05), and was associated with larger tumor size, high Lactate Dehydrogenase (LDH) and poor chemotherapy response (P < 0.05). Univariate analysis identified high IGLC1 expression (HR = 3.028, 95% CI = 1.687-5.437, P < 0.001) and elevated LDH levels (HR = 2.793, 95% CI = 1.630-4.789, P < 0.001) as significant prognostic factors for poor overall survival. Multivariate analysis confirmed both high IGLC1 expression (HR = 2.336, 95% CI = 1.214-4.495, P = 0.011) and elevated LDH levels (HR = 1.950, 95% CI = 1.062-3.579, P = 0.031) as independent predictors of poor overall survival. Functional studies revealed that IGLC1 knockdown significantly inhibited osteosarcoma cell proliferation, colony formation, migration, and invasion.</p><p><strong>Conclusion: </strong>IGLC1 is a novel independent prognostic marker that promotes tumor progression in osteosarcoma. Therapeutic strategies targeting IGLC1, such as inhibiting its expression or blocking its downstream signaling pathways, may provide innovative approaches for osteosarcoma treatment.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"931"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144149587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mengyuan Zhu, Jiehao Liao, Min Wei, Shan Huang, Junjie Xu, Qun Li, Xiaofen Pan
{"title":"A comparison of the survival outcome of paclitaxel liposome-based chemoradiotherapy with or without rhEndostatin for unresectable esophageal squamous cell carcinoma: a retrospective study.","authors":"Mengyuan Zhu, Jiehao Liao, Min Wei, Shan Huang, Junjie Xu, Qun Li, Xiaofen Pan","doi":"10.1007/s12672-025-02711-z","DOIUrl":"https://doi.org/10.1007/s12672-025-02711-z","url":null,"abstract":"<p><strong>Objectives: </strong>This study aimed to compare the survival outcomes of paclitaxel liposome-based chemoradiotherapy, with or without rhEndostatin, in patients with unresectable locally advanced esophageal squamous cell carcinoma (ESCC).</p><p><strong>Methods: </strong>Patients with ESCC treated with paclitaxel liposome-based definitive chemoradiotherapy (dCRT), with or without rhEndostatin (E), between February 2015 and June 2020 were included. Patients received induction chemotherapy followed by concurrent chemoradiotherapy, with or without rhEndostatin. The chemotherapy regimen consisted of platinum-based doublet (paclitaxel liposome + cisplatin/nedaplatin). RhEndostatin was administered at a dose of 30 mg/d from day 1 to day 5 of each chemotherapy cycle. Total radiotherapy dose was 66-68 Gy, delivered in fractions of 2.0-2.2 Gy/d. Follow-up continued until December 2023. The primary endpoints were 3-year progression-free survival (PFS) rate. Secondary endpoints included 3-year overall survival (OS) rate, objective response rate (ORR), disease control rate (DCR), and toxicity.</p><p><strong>Results: </strong>A total of 80 patients were included, with 34 in the dCRT group and 46 in the E + dCRT group. The 3-year PFS was 26.47% (95% confidence interval [CI] 13.19-41.81) in the dCRT group and 56.29% (95% CI 40.79-69.20) in the E + dCRT group (Hazard ratio (HR), 0.50; 95% CI 0.28-0.89, P = 0.012). Patients in the E + dCRT group had a superior 3-year OS compared to those in the dCRT group (80.44% [95% CI 65.77-89.30] vs. 47.06% [95% CI 29.83-62.52]; HR, 0.40; 95% CI 0.21-0.72; P = 0.003). The ORR was 91.18% in the dCRT group and 95.65% in the E + dCRT group. The most common grade 3-4 toxicities were leukopenia, neutropenia, and thrombocytopenia.</p><p><strong>Conclusion: </strong>The addition of rhEndostatin to paclitaxel liposome-based dCRT may improve clinical outcomes for patients with unresectable ESCC while maintaining manageable toxicities. However, further prospective randomized controlled studies are necessary to confirm the survival benefits of this treatment strategy.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"925"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141687","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shanshan Gu, Xu Wang, Hongxia Yang, Yaxuan Wang, Congya Yan, Xiaoting Lin, Peng Liu, Lu Liu, Li Meng, Guoyan Qi
{"title":"Identification of metabolic biomarkers and therapeutic targets in the thymoma-associated myasthenia gravis treated with methylprednisolone.","authors":"Shanshan Gu, Xu Wang, Hongxia Yang, Yaxuan Wang, Congya Yan, Xiaoting Lin, Peng Liu, Lu Liu, Li Meng, Guoyan Qi","doi":"10.1007/s12672-025-02700-2","DOIUrl":"https://doi.org/10.1007/s12672-025-02700-2","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to screen and identify metabolic biomarkers and targets for methylprednisolone treatment of thymoma with myasthenia gravis (MG) through metabolomics and network pharmacology analysis, thereby improving guidance for clinical medication and treatment.</p><p><strong>Methods: </strong>Serum from 15 patients with thymoma accompanied by severe MG was collected. Changes in serum metabolite levels before and after methylprednisolone treatment were determined using liquid chromatography-mass spectrometry (LC-MS). The raw mass spectrometry fragment information obtained was integrated and interpreted using the metabolomics data analysis software Progenesis QI v2.3. Differential metabolites were screened and identified using univariate and multivariate statistical analysis methods. Subsequently, potential targets of methylprednisolone treatment were identified through network pharmacology, and the mechanism of action of methylprednisolone in treating thymoma with MG was explored in conjunction with metabolomics. Finally, key targets and the upstream synthetic enzymes of critical metabolites identified were validated using Enzyme-Linked Immunosorbent Assay (ELISA).</p><p><strong>Results: </strong>A total of 148 differential metabolites were identified in the metabolomics study, among which key metabolites ceramide (Cer) and sphingomyelin (SM) play a significant role in cell immune regulation, inflammatory response, and tumor control. Network pharmacology analysis revealed that tumor necrosis factor (TNF) could serve as a potential target for methylprednisolone treatment of thymoma with MG. ELISA validation results showed that the key target TNF and the upstream synthetic enzymes of the key metabolites SM and Cer were all downregulated after methylprednisolone treatment, with the differences being statistically significant (P < 0.05).</p><p><strong>Conclusion: </strong>Our Study reveals that TNF could serve as a potential target for methylprednisolone treatment of Thymoma-associated MG, and Cer and SM could act as potential metabolic biomarkers to assess its treatment efficacy.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"926"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xuwen Li, Haoxi Wang, Yajian Li, Yihao Zhu, Yabo Zhai, Nianzeng Xing, Xiongjun Ye, Feiya Yang
{"title":"DNA methylation expression patterns predict outcome of clear cell renal cell carcinoma.","authors":"Xuwen Li, Haoxi Wang, Yajian Li, Yihao Zhu, Yabo Zhai, Nianzeng Xing, Xiongjun Ye, Feiya Yang","doi":"10.1007/s12672-025-02764-0","DOIUrl":"https://doi.org/10.1007/s12672-025-02764-0","url":null,"abstract":"<p><strong>Objective: </strong>To identify DNA methylation markers related to clear cell renal cell carcinoma (ccRCC) prognosis and construct a prognostic model.</p><p><strong>Methods: </strong>Methylation data from TCGA and GSE113501 dataset were analyzed. Differential analysis, univariate Cox regression, and LASSO regression were used to find survival-related CpG sites and build a risk score model. The model was evaluated by the area under the curve, and multivariate analysis determined risk factors.</p><p><strong>Results: </strong>We determined 13 CpGs that are significantly associated with prognosis through a series of regression analyses and established a risk model based on them. Patients were divided into a high-risk group and a low-risk group according to the median risk score. The results showed that there was a significant difference in the overall survival rate between the two groups (p < 0.001), and the area under the curve (AUC) of the model was greater than 0.8. Verified by the GSE113501 dataset, the model performed well in distinguishing ccRCC with different progression states. In addition, by combining methylation data with gene expression analysis, five methylation-related differentially expressed genes (LINC02541, SLAMF8, LPXN, LGALS12, EGFR) were identified, and their expression levels were significantly upregulated in tumor tissues. Multivariate analysis indicated that age, clinical stage, and methylation risk score were independent prognostic factors.</p><p><strong>Conclusion: </strong>This study confirmed that DNA methylation markers can effectively predict the progression and prognosis of clear cell renal cell carcinoma (ccRCC), providing a highly efficient and minimally invasive assessment tool for clinical practice.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"934"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144149586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Disclosing the development and focus of sequencing and omics studies in kidney neoplasm research.","authors":"Yifan Liu, Bingnan Lu, Xinyue Yang, Jinming Cui, Tianyue Yang, Haoyu Zhang, Zihui Zhao, Donghao Lyu, Yuanan Li, Yuntao Yao, Runzhi Huang, Xiuwu Pan","doi":"10.1007/s12672-025-02750-6","DOIUrl":"https://doi.org/10.1007/s12672-025-02750-6","url":null,"abstract":"<p><strong>Background: </strong>Kidney cancer is a worldwide prevalent urological malignancy and the leading cause of death. Sequencing and omics studies play a crucial role in unraveling its molecular mechanisms and diagnostic, prognostic, and therapeutic relevance. This study aims to offer a comprehensive review of the evolving trends and hotspots of sequencing and omics studies in kidney neoplasms.</p><p><strong>Methods: </strong>We conducted the retrieval of scientific publications on sequencing and omics studies in kidney neoplasms from the Web of Science Core Collection (WoSCC) on July 3, 2023. The R-based bibliometrix package, VOSviewer, and CiteSpace were utilized to conduct the holistic bibliometric analysis to obtain objective and data-driven results. A comprehensive consultation of papers was then proceeded for an in-depth review.</p><p><strong>Results: </strong>Our investigation yielded a dataset containing 1260 records from 509 sources, with 43,404 references, from 1960 to 2023. Publication and citation frequencies have been consistently growing. In country analysis, China and the USA led the research, displaying substantial collaboration. Notable contributors like TEH BT, SAUTER G, and FUTREAL PA shaped this research landscape. Key journals such as PLoS One, Cancer Research, and New England Journal of Medicine actively participated in and significantly influenced this field. Distinguished publications and references were also revealed, along with their historical citation and co-citation relationships. A panel of keywords, including RCC, biomarker, and multi-omics data were identified and clustered.</p><p><strong>Conclusion: </strong>We obtained a profound understanding of the developing trends and hotspots of research investigating sequencing and omics studies in kidney neoplasms. Specifically, we have highlighted three hotspots: \"explore molecular mechanisms of RCC pathogenesis, progression, and metastasis\", \"identify molecular biomarkers of RCC for diagnosis, prognosis, and therapeutics\", \"investigate tumor heterogeneity and tailor personalized therapeutic strategies for RCC\". Hopefully, our study will serve as a valuable reference for scientific researchers and clinical practitioners.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"928"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mingjun Ye, Shujun Chen, Chen Lu, Yangpei Wu, Jiaming Tian, Anqi Han, Jimin Zhu, Baikun Li, Qinglin Li
{"title":"Cinobufagin inhibits hepatocellular carcinoma EMT-like stemness via VEGF/VEGFR2 autocrine signaling.","authors":"Mingjun Ye, Shujun Chen, Chen Lu, Yangpei Wu, Jiaming Tian, Anqi Han, Jimin Zhu, Baikun Li, Qinglin Li","doi":"10.1007/s12672-025-02707-9","DOIUrl":"https://doi.org/10.1007/s12672-025-02707-9","url":null,"abstract":"<p><strong>Purpose: </strong>This study aimed to explore the role of the VEGFa/VEGFR2 autocrine pathway in cinobufagin-induced inhibition of hepatocellular carcinoma metastasis.</p><p><strong>Methods: </strong>A CCK-8 assay was performed to assess cell viability. Scratch healing, Transwell, and sphere formation assays were used to measure the effects of cinobufagin on cell migration, invasion, and tumor sphere formation. An immunofluorescence double staining method was used to detect the localization of VEGFa and VEGFR2. The effects of inhibiting the VEGFa/VEGFR2 autocrine pathway on Huh7 cell metastasis and the effects of the VEGFa/VEGFR2 autocrine pathway on the regulation of PI3K/AKT-dependent migration, invasion, and epithelial‒mesenchymal transition were examined through use of the VEGFR2 inhibitor apatinib and the PI3K inhibitor LY294002. The effects of cinobufagin on the VEGFa/VEGFR2 autocrine pathway and tumor metastasis were assessed in transplanted tumors.</p><p><strong>Results: </strong>Cinobufagin inhibited Huh7 cell viability, migration, invasion, and tumor sphere formation in a dose-dependent manner. In addition, colocalization between VEGFa and VEGFR2 was detected in Huh7 cells. The results revealed that apatinib treatment significantly inhibited PI3K/AKT-dependent migration, invasion, and epithelial‒mesenchymal transition. The VEGFa/VEGFR2 autocrine pathway, the PI3K/AKT pathway, and epithelial-mesenchymal transition-associated protein markers were attenuated by cinobufagin in Huh7 cells. Additionally, cinobufagin attenuated the growth of hepatocellular carcinoma tumors in the Huh7 xenograft model and significantly downregulated the VEGFa/VEGFR2 autocrine pathway, the PI3K/AKT pathway, and the epithelial‒mesenchymal transition.</p><p><strong>Conclusion: </strong>Cinobufagin may attenuate the PI3K/AKT-dependent metastatic potential of hepatocellular carcinoma by inhibiting the VEGFa/VEGFR2 autocrine pathway.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"930"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yinjiao Fei, Zhen Liu, Jinling Yuan, Lei Qiu, Yuchen Zhu, Kexin Shi, Jinyan Luo, Mengxing Wu, Weilin Xu, Shu Zhou
{"title":"A prognostic model for head and neck squamous cell carcinoma based on radiotherapy sensitivity insights from nasopharyngeal carcinoma.","authors":"Yinjiao Fei, Zhen Liu, Jinling Yuan, Lei Qiu, Yuchen Zhu, Kexin Shi, Jinyan Luo, Mengxing Wu, Weilin Xu, Shu Zhou","doi":"10.1007/s12672-025-02731-9","DOIUrl":"https://doi.org/10.1007/s12672-025-02731-9","url":null,"abstract":"<p><strong>Background: </strong>Radioresistance significantly impairs treatment efficacy and prognostic outcomes in head and neck squamous cell carcinoma (HNSCC). This study aimed to identify radiotherapy sensitivity-related genes and construct a prognostic model for HNSCC, incorporating insights from nasopharyngeal carcinoma (NPC) as a related subtype.</p><p><strong>Methods: </strong>Differentially expressed genes (DEGs) associated with radiotherapy response were identified using the GSE48501 dataset, primarily derived from NPC. Functional annotation was performed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Using the TCGA-HNSC dataset, we developed a prognostic risk model through univariate and LASSO-Cox regression analyses. The model was validated for prognostic accuracy and further analyzed for associations with immune cell infiltration, drug sensitivity, and survival outcomes using CIBERSORT, TIMER, Genomics of Drug Sensitivity in Cancer (GDSC), and nomogram analysis.</p><p><strong>Results: </strong>We identified 263 DEGs related to radiotherapy sensitivity and developed a robust prognostic model based on 8 hub genes. The model effectively stratified patients into high- and low-risk groups, with superior overall survival (OS) observed in the low-risk group. The Receiver Operating Characteristic (ROC) analysis confirmed high predictive accuracy for 1-, 3-, and 5-year OS. Immune infiltration analysis revealed reduced immune activity in the high-risk group, while drug sensitivity analysis highlighted potential therapeutic strategies. The nomogram further demonstrated excellent predictive performance.</p><p><strong>Conclusion: </strong>This study bridges insights from NPC-derived DEGs and HNSCC prognostic modeling, emphasizing radiotherapy sensitivity and integrating immune and therapeutic dimensions. The resulting model offers a novel approach to improve prognostic accuracy and guide treatment strategies for HNSCC patients.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"933"},"PeriodicalIF":2.8,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144149567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A four gene risk score model for prognosis and immune microenvironment insights in small cell lung cancer based on CAF functional-related genes.","authors":"Yunfei Chen, Yunfeng Tong, Xinyuan Ye, Yehao Yang, Hui Li, Haicheng Wu, Wanchen Zhai, Yuwei Li, Qian Zhang, Linjing Zhou, Jing Sun, Yun Fan","doi":"10.1007/s12672-025-02781-z","DOIUrl":"10.1007/s12672-025-02781-z","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is still one of the most formidable challenges in oncology. In this study, we introduce an innovative risk scoring model rooted in cancer-associated fibroblast (CAF)-related functional genes, designed to predict patient prognosis and illuminate the microenvironment of SCLC. Through Kaplan-Meier survival analysis and receiver operating characteristic (ROC) curves, our model could effectively classify patients into high- and low-risk groups, with distinct survival outcomes and remarkable predictive accuracy, which has been evidenced by the AUC values. The low-risk patients showed a more active immune environment, characterized by more infiltration of dendritic cells, natural killer cells, and higher expression of immune co-stimulation molecules. On the contrary, high-risk patients displayed an enrichment of DNA repair and glycolysis pathways associated with tumor aggressiveness and treatment resistance. These results suggest that the risk model offers a nuanced view of response to immunotherapy that may guide the identification of patients who may benefit from immunotherapy. Moreover, we also verified the function of the key gene UBE2E2 by SCLC cell line experiments. Silencing UBE2E2 results in decreased cell proliferation and migration as well as increased apoptosis, which enhances its important role in SCLC biology. In summary, our study highlights the prognostic potential of the CAF-related functional gene risk model and its implications for predicting immune microenvironment status and guiding SCLC treatment strategies.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"923"},"PeriodicalIF":2.8,"publicationDate":"2025-05-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141693","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Misbah Ullah Khan, Humaira Aslam, Jehanzeb Sohail, Ali Umar, Aman Ullah, Hayat Ullah
{"title":"Golden insights for exploring cancer: delivery, from genes to the human body using bimetallic Au/Ag nanostructures.","authors":"Misbah Ullah Khan, Humaira Aslam, Jehanzeb Sohail, Ali Umar, Aman Ullah, Hayat Ullah","doi":"10.1007/s12672-025-02714-w","DOIUrl":"10.1007/s12672-025-02714-w","url":null,"abstract":"<p><p>Sweeping contact with cancer continues to rise globally, which has led to advanced research on new treatment approaches; nanotechnology has become crucial to targeted cancer therapy. Within the intimate of nanomaterials, Au/Ag nanostructures have emerged as highly attractive because of their distinctive desirable characteristics and their prospective roles in diagnosis as well as cancer therapy. The nanostructures developed revealed remarkable biocompatibility, optically recursive alteration, and magnificently improved therapeutic effects of gold and silver in conjunction with each other. This review addresses the molecular and systemic aspects of Au/Ag nanostructures in cancer research, including the impact of nanostructures on the molecular genetic pathways and their use of systemic administration in the human organism. We explain some of the related mechanisms of action, such as photothermal therapy (PTT), and photodynamic therapy (PDT), as well as the drug delivery systems where they display potential benefits towards offering a more targeted treatment approach with fewer side effects. The latest development has shown that they have the prospect of real-time imaging and biomarker identification, and owing to this they are being viewed as a tool for individualized treatment. However, there are still some limitations: challenges of scaling up, biological safety, and bringing it to the clinic. It is therefore incumbent upon these managements to overcome these hurdles to optimize for their impact. As a result, the current findings are briefly reviewed, and the development directions are discussed to support the revolutionary role of Au/Ag nanostructures in cancer research and therapy.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"918"},"PeriodicalIF":2.8,"publicationDate":"2025-05-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yaxing Deng, Chenglin Wang, Rui Yang, Qingqiang Yang
{"title":"The role of RETN in stomach adenocarcinoma (STAD): insights from pan-cancer analysis of RNA-seq data based on the TCGA database.","authors":"Yaxing Deng, Chenglin Wang, Rui Yang, Qingqiang Yang","doi":"10.1007/s12672-025-02755-1","DOIUrl":"10.1007/s12672-025-02755-1","url":null,"abstract":"<p><strong>Objective: </strong>To analyze the expression and significance of resistin (RETN) in pan-cancer and determine the prospect of RETN as a biomarker for STAD, providing a new diagnostic and therapeutic target for STAD.</p><p><strong>Methods: </strong>Based on the RNA-seq data from the TCGA database, RETN was comprehensively analyzed from the perspective of pan-cancer. Firstly, the expression of RETN, correlation of immune infiltration, diagnostic efficacy and prognostic value in pan-cancer were investigated. Subsequently, the potential value of RETN as a biomarker for STAD was evaluated. Finally, the potential mechanisms of RETN in STAD were explored, including single-cell expression, PPI, functional enrichment analysis, single-gene correlation, gene co-expression analysis, and immune infiltration correlation.</p><p><strong>Results: </strong>RETN is upregulated in most tumors and is associated with a variety of immune cells. As a potential biomarker, RETN showed good diagnostic efficacy and prognostic value in a variety of tumors. The prediction model based on pathological stage, sex, age and RETN has a good prediction effect on the overall survival of STAD patients. Single-cell expression analysis showed that RETN was mainly expressed by DC cells and mononuclear/macrophage cells in STAD. Subsequently, RETN and related genes were analyzed from the perspective of PPI and functional enrichment, and the results showed that these genes/proteins were mainly involved in regulating hormone secretion and insulin response. Finally, RETN is also closely associated with immune cells in STAD.</p><p><strong>Conclusions: </strong>RETN is a promising biomarker related to the diagnosis and prognosis of STAD, and is expected to become a new diagnostic and therapeutic target for STAD.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"921"},"PeriodicalIF":2.8,"publicationDate":"2025-05-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141578","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}